EP3430382A1 - Proteinstrukturanalyse auf der grundlage von spektroskopie mit rotkantenanregungsverschiebung (rees) - Google Patents

Proteinstrukturanalyse auf der grundlage von spektroskopie mit rotkantenanregungsverschiebung (rees)

Info

Publication number
EP3430382A1
EP3430382A1 EP17713395.6A EP17713395A EP3430382A1 EP 3430382 A1 EP3430382 A1 EP 3430382A1 EP 17713395 A EP17713395 A EP 17713395A EP 3430382 A1 EP3430382 A1 EP 3430382A1
Authority
EP
European Patent Office
Prior art keywords
protein
indication
protein sample
characteristic
sample
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
EP17713395.6A
Other languages
English (en)
French (fr)
Other versions
EP3430382B1 (de
Inventor
Christopher Roland PUDNEY
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Bath
Original Assignee
University of Bath
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Bath filed Critical University of Bath
Publication of EP3430382A1 publication Critical patent/EP3430382A1/de
Application granted granted Critical
Publication of EP3430382B1 publication Critical patent/EP3430382B1/de
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N21/00Investigating or analysing materials by the use of optical means, i.e. using sub-millimetre waves, infrared, visible or ultraviolet light
    • G01N21/62Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light
    • G01N21/63Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light optically excited
    • G01N21/64Fluorescence; Phosphorescence
    • G01N21/6486Measuring fluorescence of biological material, e.g. DNA, RNA, cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N21/00Investigating or analysing materials by the use of optical means, i.e. using sub-millimetre waves, infrared, visible or ultraviolet light
    • G01N21/62Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light
    • G01N21/63Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light optically excited
    • G01N21/64Fluorescence; Phosphorescence
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16CCOMPUTATIONAL CHEMISTRY; CHEMOINFORMATICS; COMPUTATIONAL MATERIALS SCIENCE
    • G16C20/00Chemoinformatics, i.e. ICT specially adapted for the handling of physicochemical or structural data of chemical particles, elements, compounds or mixtures
    • G16C20/30Prediction of properties of chemical compounds, compositions or mixtures
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16CCOMPUTATIONAL CHEMISTRY; CHEMOINFORMATICS; COMPUTATIONAL MATERIALS SCIENCE
    • G16C20/00Chemoinformatics, i.e. ICT specially adapted for the handling of physicochemical or structural data of chemical particles, elements, compounds or mixtures
    • G16C20/40Searching chemical structures or physicochemical data
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N21/00Investigating or analysing materials by the use of optical means, i.e. using sub-millimetre waves, infrared, visible or ultraviolet light
    • G01N21/62Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light
    • G01N21/63Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light optically excited
    • G01N21/64Fluorescence; Phosphorescence
    • G01N2021/6417Spectrofluorimetric devices
    • G01N2021/6419Excitation at two or more wavelengths
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N21/00Investigating or analysing materials by the use of optical means, i.e. using sub-millimetre waves, infrared, visible or ultraviolet light
    • G01N21/62Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light
    • G01N21/63Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light optically excited
    • G01N21/64Fluorescence; Phosphorescence
    • G01N2021/6417Spectrofluorimetric devices
    • G01N2021/6421Measuring at two or more wavelengths
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N21/00Investigating or analysing materials by the use of optical means, i.e. using sub-millimetre waves, infrared, visible or ultraviolet light
    • G01N21/62Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light
    • G01N21/63Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light optically excited
    • G01N21/64Fluorescence; Phosphorescence
    • G01N21/6428Measuring fluorescence of fluorescent products of reactions or of fluorochrome labelled reactive substances, e.g. measuring quenching effects, using measuring "optrodes"
    • G01N2021/6439Measuring fluorescence of fluorescent products of reactions or of fluorochrome labelled reactive substances, e.g. measuring quenching effects, using measuring "optrodes" with indicators, stains, dyes, tags, labels, marks

Definitions

  • aspects and embodiments relate to a method, a computer program product and protein structure analysis unit configured to perform the method. Further aspects and embodiments provide protein structure analysis apparatus including a computer program product or protein structure analysis unit configured to perform the method and uses thereof.
  • Proteins are polypeptides formed from sequences of monomer amino acids. Protein structure determines protein function. Proteins may fold into one or more specific spatial conformations driven by various non-covalent interactions, for example, hydrogen bonding, ionic interactions, Van der Waals forces, and hydrophobic packing. Understanding the functionality of a protein at a molecular level may be assisted if it is possible to determine its three-dimensional structure.
  • proteins are thermodynamically unstable molecules and may be considered to be intrinsically flexible, dynamic molecules able to a sample a range of conformational states.
  • folded and unfolded states of a protein may differ by only the energy of a small number of hydrogen bonds.
  • conformational plasticity of a protein is an important feature of ligand binding and enzyme activity.
  • protein flexibility may also be thermodynamically unstable. That is to say, small changes in molecular environment or protein interactions may significantly alter the normal flexibility of a protein.
  • proteins such as antibodies
  • the lability of protein dynamics can be a major barrier to research, manufacture and storage of such proteins.
  • a first aspect provides a method comprising: receiving a fluorescence emission spectrum generated by a protein sample at a first excitation wavelength, the protein sample being configured to exhibit fluorescence in dependence upon its conformational state; evaluating, from the fluorescence emission spectrum, an indication characteristic of a fluorescence response of the protein sample at the first excitation wavelength; repeating the receiving and evaluating steps in relation to a plurality of fluorescence emission spectra, each fluorescence spectrum generated by the protein sample at a different excitation wavelength to the first excitation wavelength; generating a non-linear relationship between excitation wavelength and indication characteristic of fluorescence response of the protein sample, the non-linear relationship comprising an indication of at least one characteristic of the
  • proteins in general and bio-pharmaceutical proteins in particular may have a vast range of commercial, academic and therapeutic applications and that their proper function is often based on protein structure.
  • protein function may depend upon protein flexibility, for example, for antibody recognition of epitopes.
  • an analytical tool which can offer a means to detect when proteins, for example, bio-pharmaceuticals, may have lost their normal conformation and/ or flexibility and/ or begun aggregating may have significant benefits.
  • Such an analytical tool may be used within industrial manufacturing processes in relation to, for example, QC of proteins - such as antibody batches - and by formulation scientists in order to determine appropriate solvents and buffer ions and the like in which to store proteins, thus creating a stable protein sample.
  • end users may use an appropriate analytical tool provided by aspects and embodiments to check, for example, that off the shelf purchased proteins, including antibodies, are still fit for purpose.
  • embodiments may provide a simple and robust analytical technique that can be used without a need for highly specialized equipment, on a broad range of proteins, and in a variety of settings. Aspects recognise that it may be possible to use an optical phenomenon to analyse protein structure, and that by observing the characteristics of the optical phenomenon displayed by a protein sample comprising a protein, solvent and buffer, it is possible to determine a fingerprint or characterisation of that protein sample. Aspects and embodiments recognise that the characteristics of the optical phenomenon displayed by a protein sample comprising a protein, solvent and buffer map to particular features of protein structure. Aspects and embodiments may provide a mechanism to characterise a protein sample structure numerically, each numerical characteristic providing an indication of a physical characteristic of the structure of a protein within a protein sample.
  • such numerical analysis may allow for comparison of a protein sample to an "ideal" (for example, a control) protein sample, thus providing a means to assess changes in structure of a protein within a protein sample.
  • Detected changes in structure may offer an indication of the extent to which the protein in the protein sample may provide desired protein function.
  • aspects and embodiments recognise that some protein samples may, on excitation with appropriate radiation, emit radiation and that emission may comprise a fluorescence emission spectrum. Aspects and embodiments recognise that, if a protein sample emits a fluorescence spectrum in the event of appropriate stimulation, "edge shifts" in the fluorescence spectra obtained from protein samples may contain information regarding the structure of the protein. Aspects recognise that use of fluorescence phenomena to analyse the structure of a protein sample may allow for protein testing which is extremely rapid, low cost, easy to perform and information rich. Furthermore, such techniques may be non-destructive to the protein sample under test.
  • some aspects and embodiments may use an optical phenomenon called red edge excitation shift (REES).
  • REES red edge excitation shift
  • the REES effect occurs as a result of the fluorescence of molecules and may provide a mechanism to understand both inherent protein sample structure together with how proteins may interact with their surrounding environment. Aspects and embodiments recognise that it is possible to use
  • fluorescence effects exhibited by proteins such as the REES effect, to accurately report on general characteristics of a protein structure and detect changes in protein flexibility if comparing between batches of a protein or comparing aged protein samples to a fresh protein sample.
  • Some aspects and embodiments recognize emission arising from, for example, a single Trp residue may give a significant REES effect in a protein when in folded and molten globule like states, but that the REES effect may disappear or be rather reduced in unfolded states.
  • Some aspects and embodiments recognize that edge shift effects, such as the REES effect, may offer a mechanism to probe proteins with a high degree of molecular flexibility resulting in an ensemble of solvent environments around, for example, a Trp probe. In relation to proteins, such an ensemble may arise from a broad equilibrium of conformational states, such as in molten globule intermediates of highly flexible or dynamic proteins.
  • Typical analysis of fluorescence edge shift effects exhibited by a protein is such that the relationship is assumed to follow a simple linear relationship between applied excitation and resulting fluorescence emission spectra.
  • a simple linear equation is often fitted to experimentally obtained REES data.
  • Aspects and embodiments recognise that such a linear fit is poor.
  • the curvature in a plot of experimental data may be explained by the underlying physical phenomena and the structure of the protein within the studied protein sample.
  • Some aspects and embodiments quantify the REES effect by fitting the REES data to a distribution function that captures the innate curvature in REES data. From the fitted distribution function it is possible to extract values for, for example, the relative magnitude of the REES effect (A re i) and the centre of spectral mass (CSM 0 ); and aspects and embodiments recognise that those values can be related to inherent structural characteristics of the protein sample being tested. Furthermore, aspects and embodiments recognise that the extracted values may also be independent of excitation wavelength and thus offer a means to characterise or "fingerprint" a protein sample.
  • a first aspect may provide a method which comprises: receiving a fluorescence emission spectrum generated by a protein sample at a first excitation wavelength, the protein sample being configured to exhibit fluorescence in dependence upon its conformational state. It will be appreciated that not all protein samples may exhibit edge shift fluorescence effects. Accordingly, the first aspect may provide information in relation to proteins which display intrinsic fluorescence as a result of naturally occurring fluorophores within the protein structure, or which have been appropriately configured with extrinsic fluorophores at appropriate sites within the protein structure. In order to obtain useful fluorescence spectra, it will be appreciated that it may be necessary to stimulate or excite a protein sample using light or other radiation having an appropriately selected energy (wavelength) range. In particular, the excitation wavelengths may be chosen having regard to an expected fluorescence response of a given protein sample.
  • the method may comprise: evaluating, from the fluorescence emission spectrum, an indication characteristic of a fluorescence response of the protein sample at the first excitation wavelength.
  • the indication characteristic of the fluorescence response of the protein sample may comprise an indication of the wavelength at which the emission peak occurs, or the centre of spectral mass of the emission spectrum.
  • the method may comprise: repeating the receiving and evaluating steps in relation to a plurality of fluorescence emission spectra, each fluorescence spectrum generated by the protein sample at a different excitation wavelength to the first excitation wavelength. Accordingly one protein sample, either the identical protein sample or various protein samples taken from the same source, may be retested, and a series of emission spectra relating to the same protein sample maybe used to evaluate an indication
  • the temperature and pressure of the protein sample may be controlled.
  • the intensity of the radiation causing excitation at the first excitation wavelength and/ or subsequent radiation wavelengths may be controlled, to minimise the likelihood of conformational change occurring as a result of the testing or as a result of a change in the ambient environment surrounding the protein sample.
  • the method may comprise: generating a non-linear relationship between excitation wavelength and indication characteristic of fluorescence response of the protein sample. Having obtained a plurality of fluorescence emission spectra from a protein sample, it may be possible to generate a relationship between the observed macroscopic behaviour of the protein sample system and the likely conformational state of the protein with that protein sample.
  • the non-linear relationship generated may comprise an indication of at least one characteristic of the conformational state of the protein sample, based upon correlation of the received excitation wavelengths and associated evaluated indications
  • aspects and embodiments provide a fluorescence-based method which may provide a "fingerprint" reflecting the dynamic profile and structure of a protein, for example, an antibody.
  • the fingerprint may, for example, comprise a two-dimensional
  • aspects and embodiments may provide a means to discern between individual types or classes of proteins and/or a means to discern between proteins within a single type or class. Aspects and embodiments may provide a means to discern between proteins with one or more different folds, between proteins in the same or different superfamily, between proteins in the same or different families, between proteins with different protein domains, or between different species of proteins.
  • aspects and embodiments may provide a means to discern between proteins with a high level of sequence similarity. As described herein and as illustrated in Figure 12(c), the present disclosure is effective at discerning or distinguishing between proteins that have a high degree of sequence similarity. Some aspects and embodiments provide for a fingerprint generated in relation to a protein sample to be compared to a fingerprint generated in relation to a protein with a high level of sequence similarity.
  • Such proteins can have at least 80%, 85%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95% 96%, 97%, 98%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99-5%, 99.6%, 99.7%, 99.8%, 99.9% sequence similarity.
  • Such proteins can have at least 90%, 91%, 92%, 93%, 94%, 95% 96%, 97%, 98%, 99%, 99-1%, 99-2%, 99-3%, 99-4%, 99-5%, 99-6%, 99-7%, 99-8%, 99.9% sequence similarity.
  • Such proteins can have at least 95%, 96%, 97%, 98%, 99%, 99.1%, 99.2%, 99.3%, 99.4%, 99-5%, 99.6%, 99.7%, 99.8%, 99.9% sequence similarity.
  • the terms "homology, identity or similarity” refer to the degree of sequence similarity between two polypeptides or between two nucleic acid molecules encoding the two polypeptides as compared by sequence alignment.
  • the degree of homology between two discrete nucleic acid sequences being compared is a function of the number of identical, or matching, nucleotides at comparable positions.
  • the percent identity may be determined by visual inspection and mathematical calculation. Alternatively, the percent identity of two nucleic acid sequences may be determined by comparing sequence information using a computer program such as - ClustalW, BLAST, FASTA or Smith- Waterman.
  • the percentage identity for two sequences may take different values depending on: (i) the method used to align the sequences, for example, ClustalW, BLAST, FASTA, Smith-Waterman (implemented in different programs), or structural alignment from 3D comparison; and (ii) the parameters used by the alignment method, for example, local vs global alignment, the pair-score matrix used (for example, BLOSUM62, PAM250, Gonnet etc.), and gap-penalty, for example, functional form and constants. Having made the alignment, there are different ways of calculating percentage identity between the two sequences.
  • percentage identity is also strongly length dependent. Therefore, the shorter a pair of sequences is, the higher the sequence identity one may expect to occur by chance.
  • the popular multiple alignment program ClustalW Nucleic Acids Research (1994) 22, 4673-4680; Nucleic Acids Research (1997), 24, 4876-4882) is a suitable way for generating multiple alignments of polypeptides or polynucleotides.
  • a variant means a substantially similar sequence.
  • a variant can have a similar function or substantially similar function as a wild-type sequence.
  • a similar function can be at least about 50%, 60%, 70%, 80% or 90% of wild-type enzyme function or at least about 90%, 95%, 96%, 97%, 98% or 99% of wild-type enzyme function.
  • the variants can have one or more mutations that result in the enzyme having a modulated level of activity as compared to the wild-type polypeptide.
  • the variants can be mutant proteins produced by introducing any type of alterations (for example, insertions, deletions, or substitutions of amino acids; changes in glycosylation states; changes that affect refolding or isomerizations, three-dimensional structures, or self-association states), provided that they still have some or all of their function or activity.
  • Such variants can be produced by introducing any type of alterations (for example, insertions, deletions, or substitutions of amino acids; changes in glycosylation states; changes that affect refolding or isomerizations, three-dimensional structures, or self- association states), which can be deliberately engineered or isolated naturally.
  • Methods according to some aspects and embodiments may allow, for example, for the capture of subtle shifts away from a native protein structure, and, for example, any shift in native dynamics of a protein.
  • aspects and embodiments described combine numerical modelling, structure calculation and biophysical analysis to validate the general approach.
  • Some aspects and embodiments use the REES effect to provide a quantitative understanding of biomolecular edge shift (QUBES).
  • the QUBES method may allow REES measurements gathered in relation to a protein sample to yield a 2D spectral fingerprint.
  • Some aspects and embodiments provide for a fingerprint generated in relation to a protein sample to be compared to a reference fingerprint in a pre-gathered protein sample reference library to determine the quality of the protein sample being studied.
  • the 'quality' of the protein sample may relate to, but is not limited to, one or more of the conformational state of the protein in said protein sample, the crowding of the protein in the protein sample, the physical compactness of the protein in the protein sample; the stability of the protein in the protein sample; and/or the aggregation of the protein in the protein sample.
  • Some aspects and embodiments relate to detecting protein unfolding and/or formation of protein aggregates - including soluble protein aggregates.
  • Some aspects and embodiments relate to quantifying protein unfolding and/or formation of soluble aggregates - including soluble protein aggregates.
  • Some aspects and embodiments relate to predicting sample stability.
  • the methods described herein can have a number of advantages, including: (i) data acquisition and analysis is rapid ( ⁇ 5 mins) so can be used as part of large scale screening; (ii) it can be used with any protein which includes one or more Trp residues (most proteins); (iii) it can be used with proteins of any size and in nearly any solvation/buffer environment; (iv) the measurements can be high-throughput (96 well plate); (v) samples are not consumed; (vi) the approach requires only a low sample concentration; and (vii) a single value captures a complete set of information on the proteins conformation.
  • fluorescence effects to analyse the structure of a protein may have various advantages including, for example, that: (i) data acquisition and analysis is rapid ( ⁇ 5 mins) so can be used at a point of use; (ii) it can be used with any protein which includes one or more naturally occurring fluorescent amino acids (intrinsic fluorophores), and particularly those with Trp residues (most), or proteins having appropriately selected extrinsic fluorophores; (iii) it can be used with proteins of any size and in nearly any solvation/buffer environment; (iv) the measurements can be high-throughput; (v) samples are not consumed; and (vi) the approach typically requires only a small sample concentration.
  • conformational state of the protein sample comprises: an indication of a number of discrete conformational states of a protein in the protein sample. Accordingly, in some embodiments, the indication of at least one characteristic of the conformational state of the protein sample comprises: an indication of a free energy landscape of a protein in the protein sample. In other words, the fluorescence spectra produced by a protein sample may provide an inherent indication of the ruggedness of the free energy landscape of a protein sample.
  • conformational state of the protein sample comprises: an indication of a magnitude of curvature of the relationship between excitation wavelength and indication
  • characteristic of fluorescence response of the protein sample provides an indication of the magnitude of the fluorescence edge effect, for example, REES effect, displayed by a protein sample.
  • the magnitude of that effect may provide an inherent, relative or comparative indication of the nature, for example, number, of conformational states available to the protein in the protein sample. Accordingly, the magnitude of the observed fluorescence edge shift effect may provide an indication of the "ruggedness" of the free energy landscape of a protein in a protein sample.
  • conformational state of the protein sample comprises: an indication of a relative folded or unfolded nature of the conformational state of a protein in the protein sample.
  • the indication of at least one characteristic of the conformational state of the protein sample comprises: an indication of crowding of a protein in the protein sample.
  • conformational state of the protein sample comprises: an indication of physical compactness of a protein in the protein sample.
  • conformational state of said protein sample comprises: an indication of stability of a protein in said protein sample.
  • conformational state of said protein sample comprises: an indication of aggregation of a protein in said protein sample.
  • the indication of at least one characteristic of the conformational state of the protein sample comprises: an indication of the indication characteristic of fluorescence response which is determined to be independent over a range of excitation wavelength. Accordingly, across a wide range of excitation wavelengths, the fluorescence response of a protein sample may be substantially uniform. That base fluorescence response may provide information about the inherent, comparative or relative folded nature of a protein within a protein sample.
  • the non-linear relationship between excitation wavelength and indication characteristic of fluorescence response of the protein sample comprises: an exponential function, or a symmetric or asymmetric exponential probability function.
  • the relationship may comprise a Gaussian, skewed Gaussian, or sum of exponentials.
  • Such mathematical or numerical "models" for protein samples match the general shape of the expected physical behaviour of a protein configured to exhibit fluorescence effects.
  • the non-linear relationship between excitation wavelength and indication characteristic of fluorescence response of the protein sample comprises: a Gaussian probability distribution of the form:
  • A is the area
  • w is the full width at half- maximal (fwhm)
  • m is the mid-point
  • Ro is the y-intercept
  • m where is the excitation wavelength that gives the largest change in the fluorescence emission peak wavelength.
  • the protein sample comprises: a protein, solvent and buffer
  • the indication of at least one characteristic of the conformational state comprises an indication of the conformational state of the protein in the solvent and buffer.
  • the indication of the indication characteristic of fluorescence response which is determined to be independent over a range of excitation wavelength, and the indication of a magnitude of curvature of the relationship between excitation wavelength and indication characteristic of fluorescence response of the protein sample comprise a fingerprint characteristic of the protein sample. Accordingly, for a given protein sample, it may be possible to determine a fingerprint, or characteristic indicative of, for example, the free energy landscape or "ruggedness" of a protein sample and the "folded-ness" of a protein sample in a native form. Such a fingerprint may be generated by testing a protein sample immediately after manufacture.
  • Comparison of the fingerprint of an "identical" protein sample for example, a protein sample from that batch, some time after manufacture may offer an indication of whether that protein sample has a similar enough conformation (structure) to still function as intended at the point of manufacture.
  • the fingerprint may comprise two or more values characteristic or indicative of a characteristic of the protein sample. In other words, the fingerprint may be two dimensional, three dimensional ot multidimensional.
  • the method comprises: generating the indication of at least one characteristic of the conformational state of the protein sample for a plurality (for example, two or more) of different protein samples. Accordingly, it may be possible to generate a look up table comprising at least one "ideal" indication in relation to one or more protein samples.
  • the method comprises: generating the indication of at least one characteristic of the conformational state of the protein sample for a plurality (for example, two or more) of different protein samples that have a high degree of sequence similarity, as discussed herein.
  • the method comprises: comparing the indication of at least one characteristic of the conformational state of the protein sample with a previously generated indication of at least one characteristic of the conformational state of a further protein sample.
  • the further protein sample may be compared against determined characteristics of a protein sample, or set of protein samples, for which the indication of at least one characteristic of the conformational state of the protein sample is known, to identify the further protein sample. That is to say, the further protein sample may be compared against a look up table in relation to one or more "known" protein samples for the purposes of identification of the further protein sample.
  • the method comprises: comparing the indication of at least one characteristic of the conformational state of the protein sample with a previously generated indication of at least one characteristic of the conformational state of an identical protein sample. Accordingly, a protein sample may be compared against determined characteristics of an "ideal" (for example, a control) protein sample to determine whether it may still have a structure which is deemed fit for purpose. Such arrangements may allow a mechanism to perform non-destructive QC testing in relation to a protein sample.
  • the method comprises: comparing the indication of at least one characteristic of the conformational state of the protein sample, with an indication of at least one characteristic of the conformational state of a protein sample comprising the same protein having one or more different: concentration, solvent and buffer.
  • the method may allow formulation scientists to identify, by means of comparison between protein samples, a stable storage (solvent and buffer) for a given protein, thus allowing for increased likely usable biologic protein lifetimes.
  • the protein sample comprises: a protein including at least one intrinsic fluorophore.
  • the intrinsic fluorophore comprises: tryptophan.
  • the protein sample comprises a protein including at least one extrinsic fluorophore.
  • a third aspect provides a protein structure analysis unit comprising: reception logic configured to receive a fluorescence emission spectrum generated by a protein sample at a first excitation wavelength, the protein sample being configured to exhibit fluorescence in dependence upon its conformational state; evaluation logic configured to evaluate, from the fluorescence emission spectrum, an indication characteristic of a fluorescence response of the protein sample at the first excitation wavelength; collation logic configured to collate data generated by the reception and evaluation logic in relation to a plurality of fluorescence emission spectra, each fluorescence spectrum generated by the protein sample at a different excitation wavelength to the first excitation wavelength; the collation logic being configured to generate a non-linear relationship between excitation wavelength and indication characteristic of
  • the non-linear relationship comprising an indication of at least one characteristic of the conformational state of the protein sample, based upon correlation of the received excitation wavelengths and associated evaluated indications characteristic of the fluorescence response of the protein sample.
  • the protein structure analysis unit of the third aspect may be appropriately configured to provide and perform any of the features and methods described in more detail in relation to embodiments of the first aspect.
  • a fourth aspect may provide a protein structure analysis apparatus comprising:
  • a sample holder configured to receive a protein sample; a controllable light source operable to excite the protein sample; a detector operable to receive a fluorescence emission spectrum produced by the protein sample; and a control unit configured to communicate with the controllable light source and detector, the control unit comprising a protein structure analysis unit in accordance with the third aspect.
  • a fifth aspect may provide the use of the protein structure analysis unit according to the third or fourth aspects for determining an indication of a relative folded or unfolded nature of a protein, or for determining an indication of crowding of a protein; or for determining an indication of physical compactness of a protein, or for determining an indication of stability of a protein; or for determining an indication of aggregation of a protein; or for selecting an optimal protein formulation; or for monitoring protein stability, folding or aggregation
  • a sixth aspect may provide a method for comparing the stability of two or more protein samples comprising: providing two or more protein samples; performing the method according to the first aspect on each of said protein samples; and comparing the stability of the two or more proteins, wherein a difference in the indication of at least one characteristic of said conformational state of each of said proteins is indicative of a difference in stability.
  • a seventh aspect may provide a method for comparing the stability of two or more protein samples comprising: providing a protein sample; performing the method according to the first aspect on said protein sample; and comparing the stability of the protein against the stability of a second protein sample on which the method according to the first aspect has been carried out, wherein a difference in the indication of at least one characteristic of said conformational state of each of said proteins is indicative of a difference in stability.
  • An eighth aspect may provide a method for formulating a protein preparation with increased stability comprising: providing a protein in a protein sample; performing the method according to the first aspect on said protein sample in the presence of one more agents that modulate the stability of the protein; selecting one or more agents that increase the stability of the preparation in comparison to the stability of the
  • a ninth aspect may provide a method for monitoring protein stability, folding or aggregation over time comprising: providing a protein in a protein sample; and performing the method according to the first aspect on said protein at two or more different time points; wherein a difference between the results from the two or more different time points is indicative of a change in protein stability, conformation or aggregation of the protein over time.
  • a tenth aspect may provide a method for identifying one or more agents that modulate protein aggregation or protein folding comprising: providing a protein in a protein sample; performing the method according to the first aspect on said protein sample in the presence and absence of one or more test agents that may modulate the aggregation of folding of the protein; and selecting one or more agents that modulate the aggregation or the folding of the protein, wherein a difference in the level of aggregation or folding in the presence of the test agents is indicative that the test agent modulates protein aggregation or folding.
  • Figures la and lb illustrate how an example protein, NEMO W6, displays a significant REES effect
  • Figure la is a contour plot showing the change in structure of emission spectra with increasing ⁇ .
  • the dashed line of Figure la illustrates the change in the 3 ⁇ 4T for each emission spectrum and is provided as an aid to the eye only;
  • Figure lb illustrates schematically relative change the intensity of W6 emission (l3 ⁇ 4 shown as a dashed line to aid the eye) for increasing ⁇ ⁇
  • Figure lb (inset) illustrates schematically the corresponding change in CSM with . ⁇ together with an example calculated a linear fit
  • Figure 2 illustrates a phenomonological model which fits REES data.
  • the excitation spectrum of NEMO W6 (emission at 348 nm) is shown as a grey line (right-hand axis) across an extended wavelength range.
  • the variation in CSM with for NEMO W6 is shown as black circles (left-hand axis) and the solid line is an example curved fit line comprising a sum of two Gaussians which capture the ascending and descending limb of the experimentally obtained REES data;
  • Figure 3 illustrates graphically the effect of denaturation and crowding on NEMO REES data.
  • the REES data for native NEMO black circles
  • denatured NEMO red circles
  • crowded (20mg/ ml Sbi) NEMO are fit to an example curved fit line comprising a sum of two Gaussians which capture the ascending and descending limb of the experimentally obtained REES data.
  • the inset shows the extracted value of A from the example curved fit line, and is indicative of the curvature of the REES data
  • Figure 4 illustrates graphically that he REES effect is sensitive to pressure variation
  • Figure 4 a illustrates graphically variation in REES with increasing pressure.
  • Dashed lines are the fits to an example curved fit line comprising a sum of two Gaussians which capture the ascending and descending limb of the experimentally obtained REES data;
  • Figure 4b illustrates graphically the pressure dependence of A extracted from the fits of data shown in Figure 4a;
  • Figure 5 illustrates graphically a conceptual framework for interpretation of tryptophan REES data in proteins
  • Figure 6 illustrates graphically ligand induced conformational change occurs by altering the existing equilibrium of NEMO conformational states
  • Figure 6 a illustrates graphically a change in REES data on binding ⁇ and ⁇ - ⁇ peptides; the solid line in each instance comprises the fit to an example curved fit line comprising a sum of two Gaussians which capture the ascending and descending limb of the experimentally obtained REES data.
  • the Inset illustrates graphically the extracted relative magnitude of A from the fit;
  • Figure 6 b illustrates graphically a change in REES data on binding different poly- ubiquitin chain lengths; the solid line is the fit to an example curved fit line comprising a sum of two Gaussians which capture the ascending and descending limb of the experimentally obtained REES data.
  • the Inset illustrates graphically the extracted relative magnitude of A from the fit;
  • Figures 7a to 7d comprise background supporting experimental data:
  • Figure 7a illustrates the emission spectrum of tyrosine excited at 292 nm. The raw spectra, buffer spectra and difference spectrum are shown;
  • Figure 7b illustrates the emission spectrum of 2 ⁇ NEMO and 10 ⁇ tyrosine (from A) excited at 292 nm.
  • the total emission of Tyr is 0.5 % of the total NEMO emission;
  • Figure 7c illustrates the emission spectrum of NEMO excited at 292 nm (black line; left axis) and 310 nm (blue line; right axis).
  • the grey and cyan lines show the subtraction of the emission spectrum of 10 ⁇ Tyr from the NEMO emission spectra at 292 nm (black line; left axis) and 310 nm (blue line; right axis).
  • the grey and cyan lines show the subtraction of the emission spectrum of 10 ⁇ Tyr from the NEMO emission spectra at 292 nm (black line; left axis) and 310 nm (blue line; right axis).
  • the grey and cyan lines show the subtraction of the emission spectrum of 10 ⁇ Tyr from the NEMO emission spectra at 292 nm (black line; left axis) and 310 nm (blue line; right axis).
  • the grey and cyan lines show the subtraction of the emission spectrum of 10 ⁇ Tyr from the NEMO emission
  • Figure7d illustrates the resulting REES effect for NEMO (black) and for the Tyr subtracted spectra for 10 ⁇ Tyr (red).
  • Figure 8 illustrates that Sbi does not display a significant REES effect
  • Figure 8 a is a contour plot showing the change in the structure of the emissions spectra with increasing l ⁇ .
  • the dashed white line shows the change in the Jlfg* for each emission spectrum to aid the eye only;
  • Figure 8 b illustrates graphically the relative change the intensity of W6 emission
  • Gl3 ⁇ 4£" shown as a dashed black line to aid the eye for increasing A Ess .
  • Figure 8 b Ins et illustrates graphically the corresponding change in CSM with calculated from a linear fit
  • Figure 9 illustrates the pressure-dependnece of free Trp emission is neglibable compared to NEMO W6 and does not show a REES effect
  • Figure 9 a illustrates schematically a comparison of low and high pressure REES data for NEMO W6 and free Trp
  • Figure 9b illustrates schematically difference data showing the change in the REES effect for both NEMO W6 and free Trp ;
  • Figure 10 a illustrates schematically excitation and subsequent relaxation via fluorescent emission of a protein sample comprising a protein in a solvate including buffer ions;
  • Figure lob illustrates various emission spectra obtained from the protein sample of Figure 10a
  • Figure 10 c illustrates graphically a comparison of emission wavelength (x-axis) to excitation wavelength (y-axis) for the protein sample of Figure 10a;
  • Figure 11 illustrates schematically main component parts of apparatus configured to record optical shift phenomena in some arrangements
  • Figures 12a to 12c provide an overview of protein structure and analysis according to an arrangement which uses the REES effect as a specific reporter of mAb structure;
  • Figure 12a illustrates schematically a general structure of the fragment antigen- binding (Fab) region of a range of mAbs;
  • Figure 12b illustrates schematically example REES data for a mAb and shows and arrangement in which a Gaussian function is fitted to empirical data
  • Figure 12c illustrates schematically one example plot of parameters extracted from a Gaussian function fitted to empirical data as in Figure 12b for a series of different mAbs;
  • Figures 13A to D illustrate QUBES values for a range of mAbs when fit to an exponential function
  • Figures 14 A to D show how QUBES values can be used to reflect and differentiate between mAb unfolding and early stage aggregate formation
  • Figures 15A to E compares the sensitivity of QUBES values to DLS profiles
  • Figure 16 A to D shows how QUBES can be used to predict thermodynamic stability
  • Figure 17 shows the different detection capabilities of QUBES
  • Figure 18 - 21 are supporting Figures.
  • NMR techniques are understood to provide the best images of protein structures in terms of resolution, but NMR techniques require specialist equipment and take significant time. It is desirable to provide a structural analysis tool which is fast and can provide information relating to molecular flexibility. It is recognized that regardless of the level of protein structure being considered (quaternary, tertiary, secondary) the first thing to 'denature' is the normal flexibility of the molecule.
  • the flexibility of a protein is coded into the fold by the primary sequence and is also affected by the environment surrounding the protein, for example, solvent viscosity, salinity, pH, crowding and similar.
  • the native flexibility of a protein may break down very easily, for example, as a result of small changes in temperature, viscosity, pH, protein concentration and similar. It is recognized that protein flexibility is related to different protein functions including, for example, ligand binding, enzyme activity, cellular localisation.
  • Detecting ligand induced conformational change (folding) and the relationship to the equilibrium of protein conformational states is challenging.
  • EPR [3] single molecule (SM) [4,5].
  • Ion mobility mass spectrometry (IM-MS) [6] and NMR [7] studies can be used for this purpose with different levels of resolution.
  • Arrangements recognize that edge shifts on fluorescence emission spectra generated by a protein sample (for example, protein in an appropriate solution, together with appropriate buffer ions) may provide information regarding protein structure.
  • Experimental arrangements described further below demonstrate that one possible edge shift of use may comprise an optical phenomenon called Red Edge Excitation Shift (REES).
  • REES of a protein sample may provide unique information on protein conformational change and the equilibrium of conformational states [8].
  • test protein Any kind of test protein is contemplated for use in the present disclosure provided that it can be analysed by optical methods.
  • tryptophan a very convenient naturally occurring (intrinsic) fluorphore (tryptophan) which is very bright and responds to changes in its environment.
  • Tryptophan residues in proteins are extremely sensitive reporters of the immediate molecular environment. Trp residues can display a shift in their emission maximum with a decreasing energy of excitation, because the lower energy photons selectively excite discrete conformational states of the Trp-solvent system, the so called red edge excitation shift (REES) effect.
  • REES red edge excitation shift
  • Trp REES effect is a powerful tool that informs on the dynamic profile of proteins by reflecting the equilibrium of protein conformational states characterised by a proteins free energy landscape (FEL). This approach can therefore be developed to deliver sensitive detection of changes in a proteins dynamic profile as well as overall conformation.
  • FEL proteins free energy landscape
  • Antibodies including monoclonal antibodies (mAbs), are one example of test proteins that can be used in accordance with the present disclosure since they rely on native protein flexibility for function; and are generally structurally similar so identifying one characteristic which is slightly variable may be comparable across test antibodies to identify any trends in obtained experimental data. Furthermore, antibodies typically contain numerous Trp residues which are spectroscopically amenable and show emission changes with changes in surrounding environment.
  • the term "antibody” is used in its broadest sense and covers mAbs, polyclonal antibodies, dimers, multimers, multispecific antibodies (eg. bispecific antibodies), veneered antibodies, antibody fragments and small immune proteins (SIPs) (see, for example, Int. J. Cancer (2002) 102, 75-85).
  • An antibody can include a full-length immunoglobulin molecule or an immunologically active portion of a full-length immunoglobulin molecule, ie. a molecule that contains an antigen binding site that immunospecifically binds an antigen of a target of interest or part thereof.
  • the antibodies may be of any type - such as IgG, IgE, IgM, IgD, and IgA - and any class - such as lgGi , lgG2, lgG3, lgG4, IgAi and lgA2 - or subclass thereof.
  • the antibody may be or may be derived from murine, human, rabbit or from other species.
  • the antibody may be a complete antibody or an antibody fragment.
  • An antibody fragment refers to a portion of a full length antibody, generally the antigen binding or variable region thereof.
  • antibody fragments include, but are not limited to, Fab, Fab', F(ab')2, and Fv fragments; diabodies; linear antibodies; single domain antibodies, including dAbs, camelid VHH antibodies and the IgNAR antibodies of cartilaginous fish.
  • Antibodies and their fragments may be replaced by binding molecules based on alternative non-immunoglobulin scaffolds, peptide aptamers, nucleic acid aptamers, structured polypeptides comprising polypeptide loops subtended on a non-peptide backbone, natural receptors or domains thereof.
  • mAbs are of particular interest in the present disclosure. mAbs are specific, being directed against a single antigenic site and being directed against a single determinant on the antigen. mAbs may be prepared by the hybridoma method described in Nature (1975) 256:495, or they may be made by recombinant DNA methods or they may be isolated from phage antibody libraries as described in J. Mol. Biol. (1991 ), 222:581 - 597. The mAb may even be a "chimeric" antibody in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass.
  • the remaining chain(s) is identical with or homologous to sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (see PNAS (1994) USA, 81 :68si -6855).
  • the antibody may be a 'humanised antibody' ie. human immunoglobulins in which residues from a hypervariable region of the recipient are replaced by residues from a hypervariable region of a non-human species (see Curr. Op. Struct. Biol. (1992), 2:593- 596).
  • the antibody may be a 'veneered antibody'. This refers to non-human or humanized (eg.
  • the antibody may be a bispecific antibody which may comprise a hybrid immunoglobulin heavy chain with a first binding specificity in one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a second binding specificity) in the other arm.
  • the antibody may be a human engineered antibody.
  • the antibody may be a functionally active fragment, derivative or analogue of an antibody that immunospecifically binds to a desired antigen and which still recognises the same antigen that the antibody from which the fragment, derivative or analogue was derived.
  • Suitable fragments of antibodies may include F(ab')2 fragments (which comprise the variable region, the light chain constant region and the CHi domain of the heavy chain) and Fab fragments, heavy chain and light chain dimers of antibodies, or any minimal fragment thereof such as Fvs or single chain antibodies (SCAs), single domain antibodies (dAbs, IgNAR, VHH) or any other molecule with the same specificity as the antibody.
  • F(ab')2 fragments which comprise the variable region, the light chain constant region and the CHi domain of the heavy chain
  • Fab fragments fragments
  • heavy chain and light chain dimers of antibodies or any minimal fragment thereof such as Fvs or single chain antibodies (SCAs), single domain antibodies (dAbs, IgNAR, VHH) or any other molecule with the same specificity as the antibody.
  • Derivatives and analogues of antibodies are also contemplated and may include those that have been further modified by, for example, glycosylation, acetylation, pegylation, phosphorylation, amidation and/or derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular antibody unit or other protein. Chemical modifications may be carried out by known techniques - such as specific chemical cleavage, acetylation and/ or formylation.
  • the analogue or derivative may contain one or more unnatural amino acids.
  • the antibody may be a fusion protein of an antibody, an antibody-drug conjugate, or a functionally active fragment of an antibody , for example, in which the antibody is fused via a covalent bond (e.g., a peptide bond), at either the N-terminus or the C-terminus to an amino acid sequence of another protein (or portion thereof, such as at least 10, 20 or 50 amino acid portion of the protein) that is not the antibody.
  • the antibody or fragment thereof may be covalently linked to the other protein at the N-terminus of the constant domain.
  • Antibodies may be commercially obtained from a wide variety of known sources.
  • a variety of antibody secreting hybridoma lines are available from the American Type Culture Collection (ATCC, Manassas, Va.). A large number of antibodies against various disease targets have been deposited at the ATCC and/or have published variable region sequences.
  • the antibody is a human mAb.
  • the protein or antibody can be linked to a drug. It can be a therapeutic protein or antibody.
  • the drug can be a cytotoxic agent that inhibits or prevents the function of cells and/or causes destruction of cells. Examples of cytotoxic agents include radioactive isotopes, chemotherapeutic agents, and toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including synthetic analogues and derivatives thereof.
  • the protein for use in the present disclosure is in purified or isolated form, which includes a protein identified and separated and/ or recovered from a component of its natural environment.
  • an isolated or purified protein is substantially free of other cellular material or culture medium when produced by recombinant techniques, or substantially free of chemical precursors or other chemicals when chemically synthesized.
  • a protein that is substantially free of cellular material includes
  • the protein will be purified to greater than 90% by weight or 95% by weight as determined by the Lowry method, and most suitably, to more than 99% by weight, or to homogeneity by SDS-PAGE under reducing or non-reducing conditions using Coomassie blue or, preferably, silver stain.
  • purified or isolated protein will be prepared by at least one purification step.
  • the purification of the protein may include an affinity column containing agents which will bind to the protein; one or more column steps over such affinity resins; one or more steps involving hydrophobic interaction chromatography; or immunoaffinity chromatography.
  • the protein may also be expressed in a form that will facilitate purification.
  • it may be expressed as a fusion protein, such as those of maltose binding polypeptide, glutathione-5-transferase, his-tag or thioredoxin.
  • the protein may be tagged with an epitope and subsequently purified by using a specific antibody directed to such epitope.
  • One or more liquid chromatography steps - such as reverse-phase high performance liquid chromatography can be employed to further purify the protein. Some or all of the foregoing purification steps, in various combinations, can be employed to provide a substantially homogeneous protein. In certain embodiments, the same protein is used but in different formulations or at different time points or in the presence of different agents and the like.
  • One particular application of the present disclosure relates to the QC of manufactured proteins - such as bio-pharmaceutical proteins and/or recombinant proteins. All biological production systems are susceptible to genetic changes through mutation and foreign genes inserted into host cells may exhibit increased genetic instability. Proteins expressed in foreign hosts may deviate structurally, from their original, originally intended or natural counterparts.
  • tests performed at the protein level using the aspects and embodiments of the present disclosure can be used to test or compare the protein product that is produced.
  • the protein product can be compared to known (for example, control) protein products to determine the degree of variance therefrom.
  • the protein product can be compared to a fingerprint for a known (for example, control) protein product to determine the degree of variance therefrom.
  • the protein product maybe compared to a look up table comprising at least one "ideal" indication in relation to one or more protein samples to determine the degree of variance therefrom.
  • the protein product may be compared over time.
  • the aspects and embodiments of the present disclosure can be carried out at various stages or times of the manufacturing process to monitor the stability/quality of the protein product before and/ or during and/ or at the end of production.
  • the aspects and embodiments of the present disclosure can be carried out at the end of production to determine the impact of storage conditions/time on the protein.
  • Other applications of aspects and embodiments are envisaged, including, for example, testing the design and/or re-design of proteins - such as bio-pharmaceutical proteins and/or recombinant proteins.
  • the design and/or re-design may involve the use of genetic changes through mutation which can alter protein conformation/ stability and/or protein aggregation.
  • the design and/or re-design may involve the attachment of another molecule to the protein - such as a drug - which can affect protein conformation/stability and/or aggregation.
  • Tests performed at the protein level using the aspects and embodiments of the present invention can be used to determine the impact of the design and/ or re-design on the protein. If it is determined that the design and/or re-design of the protein negatively impacts the protein then further changes can be made to optimise the design and/or re-design of the protein.
  • Suitable acid addition salts are formed from acids which form non-toxic salts and include the hydrochloride, hydrobromide, hydroiodide, nitrate, sulphate, bisulphate, phosphate,
  • suitable pharmaceutically acceptable base addition salts can be formed from bases which form non-toxic salts and include the aluminium, calcium, lithium, magnesium, potassium, sodium, zinc, and pharmaceutically-active amines such as diethanolamine, salts.
  • the methods described herein can be used to decipher if the formulation of a protein into a composition (for example, a pharmaceutical composition) such as by mixing with one or more formulating agents (for example, a suitable carrier, diluent and/or excipient or the like) - has an impact on the quality, stability or conformation of the protein.
  • a composition for example, a pharmaceutical composition
  • formulating agents for example, a suitable carrier, diluent and/or excipient or the like
  • the formulation will maintain or modulate ⁇ eg. increase) the quality, stability or conformation of the protein.
  • the formulation will decrease aggregation or the likelihood of aggregation of the protein.
  • the composition may be for human or animal usage in human and veterinary medicine and will typically comprise any one or more of a pharmaceutically acceptable diluent, carrier, or excipient.
  • Carriers or diluents for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington's Pharmaceutical Sciences, Mack Publishing Co. (A. R. Gennaro edit. 1985).
  • the choice of pharmaceutical carrier, excipient or diluent can be selected with regard to the intended route of administration and standard pharmaceutical practice.
  • the pharmaceutical compositions may comprise as - or in addition to - the carrier, excipient or diluent any suitable binder(s), lubricant(s), suspending agent(s), coating agent(s), solubilising agent(s).
  • carriers include lactose, starch, glucose, methyl cellulose, magnesium stearate, mannitol, sorbitol and the like.
  • Examples of diluents include ethanol, glycerol and water.
  • binders include starch, gelatin, natural sugars such as glucose, anhydrous lactose, free-flow lactose, beta-lactose, corn sweeteners, natural and synthetic gums, such as acacia, tragacanth or sodium alginate, carboxymethyl cellulose and
  • lubricants include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
  • Preservatives, stabilizers, dyes and even flavoring agents may be provided in the pharmaceutical composition.
  • preservatives include sodium benzoate, sorbic acid and esters of p-hydroxybenzoic acid.
  • Antioxidants and suspending agents may be also used. There may be different composition/formulation requirements dependent on the intended mode of delivery.
  • the composition may be used in combination with a cyclodextrin or a derivative thereof. Cyclodextrins are known to form inclusion and non-inclusion complexes with drug molecules.
  • Formation of a drug- cyclodextrin complex may modify the solubility, dissolution rate, bioavailability and/or stability property of a drug molecule.
  • Drug-cyclodextrin complexes are generally useful for most dosage forms and administration routes.
  • the cyclodextrin may be used as an auxiliary additive, e.g. as a carrier, diluent or solubiliser.
  • auxiliary additive e.g. as a carrier, diluent or solubiliser.
  • Other factors that impact a formulation include buffer composition, pH, salt concentration and the like.
  • a further aspect relates to a method for formulating a protein preparation
  • a method for formulating a protein preparation comprising: providing a protein in a protein sample; performing the method according to the present disclosure on said protein in the presence of one more formulating agents; selecting one or more agents that maintain or modulate (eg. increase) the quality, stability or conformation of the protein preparation in comparison to the quality, stability or conformation of the protein preparation in the absence of the one or more formulating agents; and optionally formulating the protein preparation.
  • a still further aspect relates to a method for formulating a protein preparation
  • a method for formulating a protein preparation comprising: providing a protein in a protein sample; performing the method according to the present disclosure on said protein in the presence of one more formulating agents; selecting one or more agents that decreases aggregation or the likelihood of aggregation of the protein preparation in comparison to the aggregation or the likelihood of aggregation of the protein preparation in the absence of the one or more formulating agents; and optionally formulating the protein preparation.
  • a still further aspect relates to a method for selecting an optimal protein formulation comprising: providing batches of a protein containing different formulating agents; performing the method according to the present disclosure on said batches of protein in the presence of one more formulating agents; selecting one or more formulating agents that maintains or modulate (eg. increase) the quality, stability or conformation of the protein in comparison to the quality, stability or conformation of the protein in the absence of the one or more formulating agents; and optionally formulating the protein preparation.
  • a still further aspect relates to a method for selecting an optimal protein formulation comprising: providing batches of a protein containing different formulating agents; performing the method according to the present disclosure on said batches of protein in the presence of one more formulating agents; selecting one or more formulating agents that decreases aggregation or the likelihood of aggregation of the protein in comparison to the aggregation or the likelihood of aggregation of the protein in the absence of the one or more formulating agents; and optionally formulating the protein preparation.
  • the present disclosure can also be used to test proteins over time - such as in batch testing of bio-pharmaceuticals and the like. Proteins will often be stored for an amount of time post manufacture and before use and they often need testing on one or more occasions to monitor their activity. Over time, it is common for proteins to lose activity or come out of solution and/or aggregate.
  • a method for monitoring protein confirmation, stability and/or folding/unfolding over time comprising providing a protein in a protein sample; and performing the method according to the present disclosure on said protein at two or more different time points; wherein a difference between the results from the two or more different time points is indicative of a change in the activity of the protein over time.
  • Other applications of aspects and embodiments include investigating protein aggregation. For example, protein aggregation that may occur during storage or the role of protein aggregation in proteins involved in disease - such as protein aggregation in amyloidogenic proteins involved in neurodegenerative disease - can be investigated.
  • aspects and embodiments described herein may deliver novel ways of monitoring or detecting protein aggregation that may occur during storage or protein aggregation that can occur in diseases and/ or offering insight into the formation of these potentially toxic species.
  • aspects and embodiments also relate to identifying agents that can modulate (for example, decrease) protein aggregation - such as during storage or in disease.
  • aspects and embodiments therefore also relate to methods for identifying one or more test agents that modulate protein aggregation or protein folding.
  • the term "agent” may refer to a single entity or a combination of entities. The agent maybe an organic compound or other chemical.
  • the agent may be a compound, which is obtainable from or produced by any suitable source, whether natural or artificial.
  • the agent may be an amino acid molecule, a polypeptide, or a chemical derivative thereof, or a combination thereof.
  • the agent may even be a polynucleotide molecule - which maybe a sense or an anti-sense molecule.
  • the agent may even be an antibody or a part or parts thereof.
  • the agent may be designed or obtained from a library of compounds, which may comprise peptides, as well as other compounds, such as small organic molecules.
  • the agent may be a natural substance, a biological macromolecule, or an extract made from biological materials such as bacteria, fungi, or animal (particularly mammalian) cells or tissues, an organic or an inorganic molecule, a synthetic agent, a semi- synthetic agent, a structural or functional mimetic, a peptide, a peptidomimetics, a derivatised agent, a peptide cleaved from a whole protein, a peptide synthesised synthetically (such as, by way of example, either using a peptide synthesizer or by recombinant techniques) or combinations thereof, a recombinant agent, an antibody, a natural or a non-natural agent, a fusion protein or equivalent thereof and mutants, derivatives or combinations thereof.
  • biological materials such as bacteria, fungi, or animal (particularly mammalian) cells or tissues
  • an organic or an inorganic molecule such as bacteria, fungi, or animal (particularly mammalian) cells or tissues
  • the agent may be an organic compound.
  • the organic compounds may comprise two or more hydrocarbyl groups.
  • hydrocarbyl group means a group comprising at least C and H and may optionally comprise one or more other suitable substituents. Examples of such substituents may include halo-, alkoxy-, nitro-, an alkyl group, a cyclic group etc.
  • substituents being a cyclic group
  • a combination of substituents may form a cyclic group.
  • the hydrocarbyl group comprises more than one C then those carbons need not necessarily be linked to each other. For example, at least two of the carbons may be linked via a suitable element or group.
  • the hydrocarbyl group may contain hetero atoms. Suitable hetero atoms will be apparent to those skilled in the art and include, for instance, sulphur, nitrogen and oxygen.
  • the agent may comprise at least one cyclic group.
  • the cyclic group maybe a polycyclic group, such as a non-fused polycyclic group.
  • the agent may comprise at least one of said cyclic groups linked to another hydrocarbyl group.
  • the agent may contain halo groups.
  • the agent may contain one or more of alkyl, alkoxy, alkenyl, alkylene and alkenylene groups - which may be unbranched- or branched-chain.
  • the agent maybe in the form of a pharmaceutically acceptable salt - such as an acid addition salt or a base salt - or a solvate thereof, including a hydrate thereof.
  • a pharmaceutically acceptable salt - such as an acid addition salt or a base salt - or a solvate thereof, including a hydrate thereof.
  • a further aspect relates to a method for identifying one or more agents that modulate protein aggregation or protein folding comprising: providing a protein in a protein sample; performing the method according to the present disclosure on said protein sample in the presence and absence of one or more test agents that may modulate the aggregation of folding of the protein; and selecting one or more agents that modulate the aggregation or the folding of the protein, wherein a difference in the level of aggregation or folding in the presence of the test agents is indicative that the test agent modulates protein aggregation or folding.
  • a still further aspect relates to a method for monitoring protein aggregation over time comprising providing a protein in a protein sample; and performing the method according to the present disclosure on said protein at two or more different time points; wherein a difference between the results from the two or more different time points is indicative of a change in the aggregation of the protein over time.
  • a still further aspect relates to a method for screening of agents that modulate protein aggregation comprising providing the protein in a sample, adding a modulating agent, performing method set out in claims 1-20 and detecting presence or absence of change in protein conformation or stability.
  • a still further aspect relates to a method for screening for one or more agents that modulate protein aggregation comprising: providing a protein in a protein sample; performing the method according to the present disclosure on said protein sample in the presence and absence of one or more test agents that may modulate the aggregation of the protein; determining the amount of aggregation in the presence of the test agents, wherein a decrease in the amount of aggregation in the presence of the test agents is indicative that the test agent modulates protein aggregation.
  • FIGS 10a to 10c illustrate generally the REES effect observed in some proteins.
  • Figure 10a illustrates schematically excitation and subsequent relaxation via fluorescent emission of a protein sample comprising a protein in a solvate including buffer ions;
  • Figure lob illustrates various emission spectra obtained from the protein sample of Figure 10a;
  • Figure 10c illustrates graphically a comparison of emission wavelength (x-axis) to excitation wavelength (y-axis) for the protein sample of Figure 10a.
  • REES is a phenomenon where low energy excitation of a fluorophore leads to a red shift in the maximum of the emission intensity, ' . This phenomenon may manifest where there are a range of discrete fluorophore solvation states and therefore potentially reflects the equilibrium of conformational states that are accessible to a protein [8,9].
  • Figure 11 illustrates schematically main component parts of apparatus configured to record fluorescence emission spectra and thus measure optical shift phenomena in some arrangements.
  • a protein sample is prepared. That protein sample typically comprises: the protein in an aqueous solution together with appropriately selected buffer ions. The protein sample is placed in a cell holder. The sample is illuminated with substantially single wavelength collimated light of a controlled, known, intensity. The sample absorbs the energy of the incident photons and, if fluorophores are present and in a physical position within the protein and the surrounding solution which allows it, the protein sample may exhibit a fluorescence emission spectrum. The emission spectrum of the protein sample may be appropriately detected by a detector and that emission spectrum passed to an analysis unit (CPU).
  • CPU analysis unit
  • the excitation of protein sample may be repeated at a plurality of excitation wavelengths, and associated fluorescence emission spectra detected and recorded. If the excitation wavelengths are chosen appropriately to match the expected fluorescence emission spectrum of the protein, it may be possible to analyse the various obtained emission spectra and obtain information relating to any edge shift effects including, for example, the REES effect exhibited by a protein sample.
  • One arrangement may provide an experimental method comprising: receiving a fluorescence emission spectrum generated by a protein sample at a first excitation wavelength, said protein sample being configured to exhibit fluorescence in
  • Figures 12a to 12c provide an overview of protein structure and analysis according to an arrangement which uses the REES effect as a specific reporter of mAb structure.
  • Figure 12a illustrates schematically a general structure of the fragment antigen-binding (Fab) region of a range of mAbs and such a protein maybe tested in apparatus such as that shown and described in relation to Figure 11.
  • Fab fragment antigen-binding
  • Figure 12b illustrates schematically example REES data for a mAb such as that shown in Figure 12a.
  • Figure 12b shows an arrangement in which a Gaussian function is fitted to empirical data.
  • Figure 12c illustrates schematically one example plot of parameters extracted from a Gaussian function fitted to empirical data as in Figure 12b for a series of different mAbs.
  • A is the area
  • w is the full width at half- maximal (fwhm)
  • m is the mid-point
  • CSMo is the y-intercept
  • m where is the excitation wavelength that gives the largest change in the emission peak wavelength.
  • Equation 10 The empirically obtained REES data displays a good fit to Equation 10 and illustrate the low energy excitation at the red edge of the protein absorption spectrum.
  • the fit of Equation 10 to the experimental data is good and captures expected relationships as described in more detail in relation to specific experiments set out in more detail below.
  • the magnitude of A extracted from fitting Equation 10 to a plot of excitation wavelength versus gJ ⁇ can be used as a qualitative comparator to assess changes in the extent of the observed REES phenomenon.
  • the magnitude of R 0 should represent the minimum value, in the absence of the REES effect. This magnitude is commonly used to reflect the degree of solvent exposure of, for example, Trp residues in a protein sample and may, for example, be used as a metric of protein folded/unfolded states. Fitting experimental data to a suitable numerical model does not recapitulate the absorbance/excitation spectrum of a fluorophore yet may still provide a physically meaningful probe.
  • Non-denaturing pressure perturbation is an excellent tool for testing if REES is sensitive to changes in the number of discrete equilibrium conformational states of a protein sample, since changing pressure acts to perturb a pre-existing equilibrium of structural states characterized by different energy minima on the protein FEL [33]. Pressure has been used as a perturbation in a number of studies [34-36]. It will be understood that pressure, unlike temperature, does not alter the internal energy of the system, which might otherwise be a confounding factor for REES measurements. The observation of a significant pressure dependence on the REES effect itself is therefore be powerful evidence that REES is sensitive to the equilibrium of protein
  • the pressure dependence data can be adequately fit to a simple, phenomenological model which implies a single transition between two states with changing hydrostatic pressure as used previously [23,38].
  • the key finding from the pressure data in the present context is that the REES effect itself is pressure-dependent with the magnitude of A increasing with pressure. Given that pressure acts to perturb the equilibrium of conformational states, this observation suggests that the magnitude of A extracted from Equation 10 is sensitive to this equilibrium.
  • Protein samples subjected to denaturation, pressure changes and macromolecular crowding (protein stabilization) measurements produce empirical data which suggests that the REES effect as modelled by Equation 10, is sensitive to changes in the equilibrium of conformational states, for example, by unfolding (chemical
  • Equation 10 a conceptual framework for interpreting protein REES data using Equation 10, as shown in Figure 5.
  • some aspects and embodiments recognise the information content that arises from, for example, the curvature of the REES data.
  • the changing curvature of the REES data reflected by the magnitude of A from in Equation 10, describes the progression to a larger or smaller number of discrete conformational states, a rugged or flat FEL, respectively.
  • the intercept with the y-axis, Ro can be used as an indicator of whether the protein sample tends towards a folded or unfolded state, in exactly the same way as might be expected from a traditional analysis of Trp emission in protein folding studies, but with the added benefit that the numerical model of Equation 10 as fitted to empirical data takes account of cases where the peak maximum of the emission band shifts with excitation wavelength.
  • the terms "folded” and "unfolded” used on the context of the framework of Figure 5 also encompass more minor conformational changes. So, for highly flexible and dynamic proteins, a blue shifted CSM may simply reflect a more compact conformational state (as suggested is the case from the crowding experiment results shown in Figure 5), without requiring a large scale folding event.
  • NF-KB essential modulator is the key regulatory element in the NF- ⁇ signaling pathway, controlling much of the nervous and immune system [18].
  • NEMO regulates the activity of a kinase, ⁇ kinase- ⁇ ( ⁇ - ⁇ ), which has a diverse range of kinase, ⁇ kinase- ⁇ ( ⁇ - ⁇ ), which has a diverse range of kinase, ⁇ kinase- ⁇ ( ⁇ - ⁇ ), which has a diverse range of
  • phosphorylation targets for example, ⁇ [19], huntingtin [20], and insulin receptor 1 (IRS-i) [21].
  • NEMO putatively ensures the specificity of ⁇ - ⁇ for ⁇ , by facilitating the recruitment of ⁇ the kinase [19]. Subsequent proteosomal degradation of ⁇ then allows the NF- ⁇ complex to enter the nucleus and induce expression of proinflammatory and anti-apoptotic genes.
  • NEMO putatively ensures the specificity of ⁇ - ⁇ for ⁇ , by facilitating the recruitment of ⁇ the kinase [19]. Subsequent proteosomal degradation of ⁇ then allows the NF- ⁇ complex to enter the nucleus and induce expression of proinflammatory and anti-apoptotic genes.
  • NEMO Despite the importance of NEMO to normal human health and disease associated processes, remarkably little is known about the molecular mechanism of action of NEMO and, in particular how NEMO is able to show specificity to a very large number of interaction
  • NEMO is a flexible protein that undergoes ligand specific conformational change, and have hypothesized that the non-ligand bound protein adopts a broad equilibrium of conformational states [23].
  • the emission of NEMO's single intrinsic Trp residue (W6) has been used as a spectroscopic reporter of ligand- induced NEMO conformational change [23].
  • REES spectroscopy is described to inform on the structural and mechanistic determinants of NEMO-ligand binding.
  • a new framework for the quantitative interpretation of steady-state REES data is described, validated by folding and pressure perturbation studies. This then allows us to use the REES data quantitatively for a series of NEMO-ligand bound states.
  • a putative model for the molecular mechanism of NEMO's functional interactions, structure and how this connects to the NEMO free energy landscape and conformational change is developed.
  • Figure l shows the variation in emission spectrum (Figure lA and iB) and center of spectral mass (CSM) ( Figure iB) of NEMO W6 versus excitation wavelength.
  • CSM center of spectral mass
  • Figure iB the magnitude of the steady-state REES phenomenon is reported as the simple difference in CSM or From Figure l, it is observed that a significant red shift in the emission spectra, with an increase in CSM of 17.7 nm and an increase in of 15 nm from Aex 292-310 nm.
  • the data are treated by the linear function: Where R is the REES magnitude expressed as the change in CSM per nm of the excitation wavelength, Aex.
  • Pan et al have previously demonstrated that even solvent exposed Trp residues can display a broad range of values, arising from different Trp rotamers [26] and calculate a range 344 - 365 nm from molecular dynamics simulations for a range of cyclic peptides. This would seem consistent with the data that show a similar range of values. Further, Maglia et al find a significant REES effect in a single Trp variant of DD-carboxypeptidase attributable to at least three different Trp rotamers [27]. These rotamers may be stabilized through differing H-bonding to the Trp amide carbonyl [28].
  • Trp rotamers As such it is difficult to directly separate the contribution of differing Trp rotamers and larger scale conformational heterogeneity of the peptide backbone to the REES signal. Below, the contribution of different Trp rotamers to the NEMO REES signal in detail is investigated.
  • the REES signal is monitored from a single Tyr containing fragment of Staphylococcus aureus complement evasion second immunoglobulin-binding protein Sbi domain III and IV that, like NEMO, is composed of a significant fraction of structurally disordered content [29].
  • Figure 8 A significant REES signal arising from Tyr emission is not found as is expected because of the symmetrical nature of the Tyr ring system. Together, these data demonstrate that the signal monitored, as expected, arises essentially entirely from NEMO's single intrinsic Trp. Estab lis hing a q ua ntitativ e a na ly s is of REES data .
  • the REES data in Figure 2 can be best represented by a Gaussian probability distribution of the form:
  • A is the area
  • w is the full width at half- maximal (fwhm)
  • m is the mid-point
  • Ro is the y-intercept and Tii— where ⁇ 3 ⁇ 4f3 ⁇ 4 is the excitation wavelength that gives the largest change in the emission peak wavelength.
  • the data in Figure 2 are fit to Eq 2 and illustrate the low energy excitation at the red edge of the protein absorption spectrum. The fit of Eq 2 to the experimental data is excellent and captures the expected relationship described above.
  • the magnitude of A extracted from fitting Eq 2 to a plot of excitation wavelength versus can be used as a qualitative comparator to assess changes in the extent of the REES phenomenon.
  • the magnitude of R 0 should represent the minimum j3 ⁇ 4* value, in the absence of the REES effect. This magnitude is commonly used to reflect the degree of solvent exposure of Trp residues and can be used as a metric of folded/unfolded states. The interpretation of these values is described in more detail below. This simple model clearly neglects a range of contributing factors such as the proportion of excited molecules, the small contribution from changing excitation energy at different excitation wavelengths, the number of discrete conformational states that are photo-selectable and the contribution from dipole rotation in the excited electronic state.
  • Eq 2 unlike Eq 1, is based on a realistic physical rationale and provides a means to extract meaningful quantitative data from the full range of REES data.
  • Other distribution functions can and have been used in relation to extracting information from spectral features, including Lorentz and log-normal distributions [30-31].
  • NEMO ⁇ 1.23 ⁇ 0.0.1 362.8 ⁇ 0.25 0.93 ⁇ 0.1 1 a Values are relative to the extracted value for NEMO alone. Typically one does not expect to observe a significant REES effect with denatured protein since the peptide backbone will become fully solvent accessible. In this case, the solvent relaxation will be very rapid and no REES effect observed, effectively because there is one discrete solvation state (fully solvated).
  • NEMO is incubated in 6M guanidine to denature the protein and the corresponding change in the center of spectral mass (CSM) versus Aex is shown in Figure 3.
  • CSM center of spectral mass
  • the denatured NEMO shows a dramatically reduced REES effect.
  • Macromolecular crowding is a key feature of the intracellular milieu arising from the very high concentrations of other species in the cytosol. Crowding reduces the available solvent for other molecules in solution through the excluded volume effect, which effectively increase the concentration of, for example, protein and restricts the accessible conformational states through hard-core repulsions between the crowding agent and the protein. Macromolecular crowding can alter protein conformation, typically inducing folding and stabilization [32]. Crowding therefore potentially provides a means to explore the contrasting physical effect of denaturation on the REES effect, where the protein is more folded/stabilized. A protein has been used as the crowding agent, Sbi (reported above), instead of a synthetic crowding agent since protein is much more consistent with the intracellular environment.
  • Sbi lacks any intrinsic Trp residues and does not give rise to a REES signal that would convolute the NEMO signal (Figure 7).
  • Incubating NEMO with a high concentration of Sbi (20 mg/ ml) gives a dramatic change to the both the absolute Trp emission and also the REES effect as shown in Figure 3.
  • K 0 83.13 cm3 mol 1 bar
  • K 0 is the equilibrium constant for the change in the relative population of the ith conformational state represented by the magnitude of A from Eq 4, extrapolated to o bar
  • AV A is the apparent difference in the volume associated with this equilibrium transition.
  • the REES signal may therefore be reflective of the broader equilibrium of NEMO conformational states at sites distal to W6. That is, these two values are not necessarily comparable, reflecting different aspects of the W6 molecular environment.
  • the key finding from the pressure data in the present context is that the REES effect itself is pressure-dependent with the magnitude of A increasing with pressure. Given that pressure acts to perturb the equilibrium of conformational states, this observation suggests that the magnitude of A extracted from Eq 2 is extremelyly sensitive to this equilibrium.
  • the changing curvature of the REES data reflected by the magnitude of A from Eq 2, describes the progression to a larger or smaller number of discrete conformational states, a rugged or flat FEL, respectively.
  • the intercept with the y-axis, R 0 describes whether the protein tends towards a folded or unfolded state, in exactly the same way as per the normal analysis of Trp emission in protein folding studies, but with the added benefit that it takes account of cases where the peak maximum of the emission band shifts with excitation wavelength. It is important to note that the terms folded and unfolded encompass more minor conformational changes also. So for highly flexible and dynamic proteins a blue shifted CSM may simply reflect a more compact conformational state (as suggested is the case from the crowding experiment shown in Figure 2), without requiring a large scale folding event.
  • Figure 6A shows NEMO alone and bound to either a peptide mimic of ⁇ or ⁇ - ⁇ .
  • the ⁇ peptide (termed the NEMO binding domain, NBD) contains two Trp residues and so is not suitable for the present study as the signal from the peptide would confound the analysis of the single NEMO Trp. Instead these Trp residues are replaced with a conservative amino acid, Phe, and called NBD-Phe. it is found that this modified peptide binds to NEMO, giving a decrease in W6 emission of ⁇ 50 %. From Table 1 and Figure 5A there is a significant difference in the magnitude of A depending on the ligand bound form of NEMO.
  • NEMO comprises a specific domain that non-covalently binds poly-ubiquitin. It has previously been found that binding of long chain-length 'free' Mi-linked poly-ubiquitin chains to NEMO allosterically regulates ligand affinity and potentially cellular localization based on evidence from stopped-flow and liposome binding assays [23]. Poly-ubiquitin is found as a range of chain-lengths in the cell and evidence has previously been provided from ANS binding studies that NEMO undergoes different conformational change depending on the chain-length of non-covalently bound poly- ubiquitin [23]. The REES effect on NEMO W6 is explored in the presence of both short (4-mer; Ub 4 ) and long (10-mer; Ubi 0 ) poly-ubiquitin chains, as shown in Figure 6B.
  • NEMO predominately utilizes a conformational selection model of ligand binding. That is, NEMO adopts a range of equilibrated conformational states represented by discrete energetic minima on the protein FEL. Ligand binding occurs at one of these pre-existing conformers, shifting the equilibrium towards the ligand bound population.
  • Full length human NEMO was expressed and purified essentially as described previously [23]. Purified protein was dialysed extensively into a buffer comprising 50 mM Tris-Cl pH 8.0, 50 mM NaCl and 5 mM DTT. All measurements were made in this buffer unless otherwise stated. ⁇ - ⁇ and ⁇ peptides were commercially
  • TALDFSFLQTE and DDRHDSGLDSMKD were modified such that the two native Trp residues were replaced with Phe residues.
  • Mi- linked poly-ubiquitin was purchased from Viva Bioscience. Staphylococcus aureus immune modulator protein fragment Sbi-III-rV K173A was expressed and purified essentially as described previously [44]. Protein concentration used was between 1 and 5 ⁇ . Peptide concentrations were 1 mM and poly-ubiquitin concentrations were 1 ⁇ .
  • Luminescence Spectrometer Perkin Elmer, Waltham, MA, USA
  • ⁇ 1 °C temperature regulation
  • Samples were incubated for 5 minutes at the given conditions prior to recording measurements. Measurements were performed at io°C, unless otherwise stated.
  • Excitation and emission slit widths were 5 nm except for pressure experiments where they were 10 nm. The larger slit width was required due to the optical setup of the pressure cell to ensure a low signal- to-noise for the W6 emission signal.
  • tryptophan emission was monitored from 315 to 550 nm. The excitation wavelength was subsequently increased in inm steps for a total of 19 scans.
  • ISS high-pressure cell (ISS, Champaign, UL, USA), fitted with a custom fiber optic mounting to the fluonmeter and connected to a circulating water bath for temperature regulation was used to record all high-pressure measurements.
  • tryptophan emission was monitored between 325-450 nm.
  • CSM center of spectral mass
  • GSW - Eq 4 Where fi is the measured fluorescence intensity and A em is the emission wavelength. The importance of using a consistent wavelength range when reporting CSM data is stressed, as the magnitude will be dependent on the wavelength range chosen. As such, the CSM across the emission range 325 - 450 nm or 335 - 450 nm is reported for pressure experiments.
  • the edge-shift effects for a range of mAbs have been monitored and are shown in Figure 13 A-C.
  • the combined excitation and emission spectrum is monitored for each mAb giving a high information content fluorescence data set ( Figure 13A).
  • the intensity and peak position of the emission incorporates information on (i) the number of Trp residues in the sample (ii) the degree of solvent exposure of the Trp residues 24 including arising from different rotamers (iii) energy transfer to the peptide
  • CSM 0 is the CSM value independent of the excitation wavelength, ⁇ ⁇ , determined by the amplitude, A, of an exponential with a curvature determined by R. Fitting with this function instead of a Gaussian or other probability distribution dramatically improves the speed of data processing due to the lower dimensionality of the model but still retains the key aspects of the previous model, namely quantification of the magnitude of the fluorescence edge-shift.
  • the plot of the resulting parameters yields a single three-dimensional data point ( Figure 13B, inset; the QUBES value), which is a direct quantification of the extremely complex spectral fingerprint shown in Figure 13A.
  • QUBES values for a range of therapeutic mAbs are measured from different classes including, chimeric (ximab), humanised (zumab) and human (lumab) in the same buffer system.
  • the humanised mAbs are shown in Figure 31C and all of the examples in Figure 13D.
  • Pembrolizumab Pembrolizumab, Vedolizumab and Natalizumab, which are IgG4, differing only by three residues in the hinge region and retaining the same inter- heavy chain disulphide bonds.
  • the three-dimensional structures can be considered to be essentially identical.
  • the far-UV circular dichroism spectrum and dynamic light scattering (DLS) profile of these full length mAbs is highly similar if not identical (see below) as expected for proteins with high sequence similarity and similar overall structures particularly in the percentage of secondary structure content.
  • the Fab region ( Figure 18) contains the most sequence variability and there are some small differences in the number and/or position of Trp residues for some of the mAbs in the Fab region ( Figure 18 and 18B and Table of Figure 21). However, these differences (number of residues and solvent accessible surface area) do not correlate show a correlation with the extracted QUBES values ( Figure 18C and 18D). It was previously suggested that the curvature in the REES effect captures information on the equilibrium of conformational states accessible on the FEL, which can be thought of as the proteins relative
  • the observed variance in the QUBES values for the denatured mAbs may reflect the differing extent of unfolding or aggregation for each of the samples and the specific nature of the unfolded or aggregated states. This is confirmed by monitoring the sequential unfolding of Tocilizumab and aggregation of Rituximab shown in Figure 14B and 14C, respectively.
  • the unfolding of Tocilizumab proceeds with an initial shift in the QUBES value to a larger R and smaller A value ( Figure 14B). This finding is consistent with other studies that report a smaller overall edge-shift for unfolded versus native protein since the protein is tending towards a single conformational state; unfolded.
  • Nivolumab is monitored by both QUBES and DLS and the data are shown in Figure 15A and 15B-E, respectively.
  • An increase in A and a decrease in R and CSM 0 is observed, correlated with increasing formation of soluble aggregates.
  • Both QUBES and DLS data show a significant shift at l hr incubation ( Figure 15B).
  • the QUBES values show a significant shift prior to a change in the volume data from DLS ( Figure 15B and 15C) and even where there are only subtle shifts in the intensity data (Figure 15D).
  • Intensity DLS profiles are vastly more sensitive than volume DLS profiles, but volume data are typically preferred as they can potentially be quantitative and often suffer from false positives.
  • the QUBES value shows a significant shift prior to a change in the volume of DLS profiles, the QUBES value can be used to quantify the percentage of soluble aggregates with detection that is at least as sensitive and accurate as DLS.
  • the QUBES approach should be extremely sensitive to protein secondary, tertiary and quaternary structure change. This may be because it captures information on subtle changes to the proteins equilibrium of conformational states. The data corroborate this, demonstrating that the QUBES value is sensitive to, and can quantitate, increasing fractions of unfolded or aggregated species at very high sensitivity.
  • Pertuzumab may be more stable than those predicted to be relatively flexible (high A and small R value) e.g. Nivolumab.
  • the thermal stability of three mAbs with QUBES values has been explored suggesting increasing flexibility (Pertuzumab, Vedolizumab and Nivolumab), by monitoring the formation of soluble aggregates at the same temperature for each mAb, shown in Figure 16A.
  • the QUBES value can be used in predictive manner to infer the relative thermodynamic stability of a sample.
  • the predictive power is only appropriate for the same sample, since each protein will exhibit a specific QUBES signature.
  • the mAbs have three dimensional structures that are essentially identical and so the comparison between them is valid.
  • the QUBES analysis has the advantages that: (i) data acquisition and analysis is rapid ( ⁇ 5 mins) so can be used as part of large scale screening; (ii) it can be used with any protein which includes one or more Trp residues (most proteins), (iii) using proteins of any size and in nearly any solvation/buffer environment; (iv) the measurements can be high-throughput (96 well plate); (v) samples are not consumed, (vi) the approach requires only a low sample concentration and (vii) a single QUBES value captures a complete set of information on the proteins conformation. The approach is designed to be used in a comparative fashion and is most robust and finds best utility when examining e.g. variants of buffer conditions for the same protein.
  • QUBES may find immediate utility in both the formulation and quality assurance of biopharmaceuticals, but also in academic labs more generally that seek to understand the role of protein dynamics in protein function. For example, there is presently intense scrutiny of the role of early stage protein aggregation in amyloidogenic proteins involved in neurodegenerative disease. Given the very high sensitivity of QUBES to detecting and quantifying protein soluble protein aggregates, the use of the approach will deliver novel insight into the formation of these potentially toxic species.
  • f is the measured fluorescence intensity and A em is the emission wavelength.
  • a em is the emission wavelength. The importance of using a consistent wavelength range when reporting CSM data is described, as the magnitude will be dependent on the wavelength range chosen.
  • the QUBES data are extracted by fitting the CSM versus AEX data as described in the manuscript. Data fitting and plotting was performed using OriginPro 2016 (Microcal).
  • Antibody samples unfolding and aggregation.
  • Antibodies were provided by Bath ASU and were either extensively dialysed (for urea denaturation experiments) or diluted into Tris-Cl buffered saline pH 8. All buffer components were of a spectroscopic grade.
  • Antibody denaturation was achieved by extensive dialysis into a buffered solution of 8M urea or oM urea as a control.
  • Antibody aggregation was achieved through incubation at elevated temperatures and monitored by DLS.
  • Fig 13 shows QUBES values for a range of mAbs that are correlated with differences in molecular flexibility.
  • A The QUBES value are extracted from a combined excitation- emission spectrum for protein Trp residues.
  • B The CSM versus excitation wavelength (grey data) is fit to Eq 1, to give a single data point governed by 3 parameters and this is called the QUBES value (shown in inset).
  • C QUBES values for a series of Kursab examples; Pembrolizumab (green), Vedolizumab (blue), Pertuzumab (orange), Natalizumab (yellow), Bevacizumab (Indigo), Trastuzumab (red), Trastuzumab emtansine (light blue) and Tocilizumab (light green).
  • D The QUBES values reflect the difference in intra-molecular flexibility between mAbs of the same class showing Kursabs coloured red, ximabs (Rituximab, Infliximab and Cetuximab) coloured blue and Nivolumab coloured black. The corresponding coloured planes represent the 30% confidence interval for the data sets and are to aid the eye.
  • FIG 14. The QUBES value can be used to accurately reflect and differentiate between mAb unfolding and early stage aggregate formation. QUBES values for Kursab shown in Figure 13C (red), incubated in 8M urea (red) and thermally aggregated (purple).
  • B Tocilizumab QUBES data under increasing concentrations of urea (red numbers in are [urea] (M)) titration of Urea
  • C the sensitivity of QUBES value to aggregation. The data show the titration of fully aggregated Rituximab into native protein with the data points sequentially from left to right corresponding to o, 5, 9. 13, 17 and 23 % aggregate.
  • D Numerical modelling of increasing fractions of unfolded (blue) and aggregated (brown) Tocilizumab based on fully unfolded (8M urea) and fully solubley aggregated sample QUBES values.
  • FIG. 15 Comparing the sensitivity of the QUBES value to DLS profiles.
  • A QUBES values for Nivolumab incubated at 65 °C with values recorded every 30 minutes.
  • B-E Corresponding DLS profiles showing both % intensity (solid lines) and % volume (dashed lines) readings compared to the fresh sample (black). No change in the DLS profiles was observed at 0.5 hrs.
  • thermodynamic stabilities Thermally driven aggregation of the mAbs in panel B for 3 hours [Pertuzumab (red) and Vedolizumab (green)] and [Nivolumab (blue)] at 65 °C and a concentration of 0.5 mg / ml. Arrows indicate the transition from ohrs to 3 hours incubation.
  • D Thermally driven aggregation (5 days at 37 °C) of Pemrolizumab (black) in the presence (orange) and absence of 60 mM glucose, respectively. Arrows indicate the transition from the beginning to the end of the incubation.
  • E Corresponding DLS profiles from panel D. Black solid line represents a non-aggregated mAb.
  • Figure 18 A PyMol representation of an overlay of all RosettaAntibody homology models.
  • Figure 18A The conserved tryptophans are displayed in stick representation and are highlighted in orange. It can be seen that the conserved 47 th residues on the V H chain (boxed) have been modelled at two different orientations. The other conserved tryptophan residues are modelled in the same orientation.
  • Figure 18B shows the same image, but with all (conserved and un-conserved) residues highlighted in blue. It is clear that the additional un-conserved tryptophan residues are located in a similar area of the molecule, and appear to be closer to the edge of the molecule (less-buried).
  • Figure 18C shows the correlation of the QUBES value with Trp solvent accessible surface area (SASA) in the Fab region.
  • Figure 18D shows the c orrelation of the QUBES value with the number of Trp residues in the Fab region.
  • Figure 19 shows the DLS profiles for thermally aggregated mAbs shown in Figure 14A.
  • Figure 20 shows the correlation between the changing magnitude of the QUBES value represented by the parameters A and R with increasing unfolded content (blue) and aggregated content (brown).
  • Figure 21 shows a supporting information table showing a summary of calculated parameters for mAb Fab regions.
  • Demchenko AP (1988) Red-edge-excitation fluorescence spectroscopy of single- tryptophan proteins. Eur. Biophys. J. 16, 121-129.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Immunology (AREA)
  • Physics & Mathematics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Computing Systems (AREA)
  • Theoretical Computer Science (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Investigating, Analyzing Materials By Fluorescence Or Luminescence (AREA)
EP17713395.6A 2016-03-18 2017-03-17 Analyse der struktur von proteinen mittels red-edge-excitation-shift (rees) spectroskopie Active EP3430382B1 (de)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1604640.1A GB201604640D0 (en) 2016-03-18 2016-03-18 Method and apparatus
PCT/GB2017/050743 WO2017158371A1 (en) 2016-03-18 2017-03-17 Protein structure analysis based on red-edge excitation shift (rees) spectroscopy

Publications (2)

Publication Number Publication Date
EP3430382A1 true EP3430382A1 (de) 2019-01-23
EP3430382B1 EP3430382B1 (de) 2020-05-06

Family

ID=55968544

Family Applications (1)

Application Number Title Priority Date Filing Date
EP17713395.6A Active EP3430382B1 (de) 2016-03-18 2017-03-17 Analyse der struktur von proteinen mittels red-edge-excitation-shift (rees) spectroskopie

Country Status (5)

Country Link
US (1) US10900903B2 (de)
EP (1) EP3430382B1 (de)
ES (1) ES2795997T3 (de)
GB (1) GB201604640D0 (de)
WO (1) WO2017158371A1 (de)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109101785B (zh) * 2018-07-12 2021-06-18 浙江工业大学 一种基于二级结构相似性选择策略的蛋白质结构预测方法

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5674664B2 (ja) * 2008-09-04 2015-02-25 ベックマン コールター, インコーポレイテッド パンキナーゼ活性化およびシグナリング経路の評価
US20130137127A1 (en) 2011-11-30 2013-05-30 Washington University Dating bloodstains and biological fluids with fluorescence lifetime techniques
US9188592B2 (en) 2012-10-10 2015-11-17 Oregon Health & Science University Fluorescent detection of in vitro translated protein on a solid surface
US9588129B2 (en) 2013-03-15 2017-03-07 Amira Medical Technologies Inc. Methods for analyzing blood to detect diseases associated with abnormal protein aggregation

Also Published As

Publication number Publication date
EP3430382B1 (de) 2020-05-06
US20190072491A1 (en) 2019-03-07
GB201604640D0 (en) 2016-05-04
US10900903B2 (en) 2021-01-26
ES2795997T3 (es) 2020-11-25
WO2017158371A1 (en) 2017-09-21

Similar Documents

Publication Publication Date Title
Keiderling Structure of condensed phase peptides: insights from vibrational circular dichroism and Raman optical activity techniques
Xu et al. Structure, heterogeneity and developability assessment of therapeutic antibodies
Greenfield Analysis of the kinetics of folding of proteins and peptides using circular dichroism
Thiagarajan et al. A comparison of biophysical characterization techniques in predicting monoclonal antibody stability
Razinkov et al. Accelerated formulation development of monoclonal antibodies (mAbs) and mAb-based modalities: review of methods and tools
Wang et al. Exploring weak, transient protein–protein interactions in crowded in vivo environments by in-cell nuclear magnetic resonance spectroscopy
Johnson Differential scanning calorimetry as a tool for protein folding and stability
Matheus et al. FTIR and nDSC as analytical tools for high-concentration protein formulations
Bloem et al. Ligand binding studied by 2D IR spectroscopy using the azidohomoalanine label
Hudgens et al. Interlaboratory comparison of hydrogen–deuterium exchange mass spectrometry measurements of the Fab fragment of NISTmAb
Bagchi et al. Dynamics of a myoglobin mutant enzyme: 2D IR vibrational echo experiments and simulations
Liu et al. Lys63-linked ubiquitin chain adopts multiple conformational states for specific target recognition
Shashilov et al. 2D correlation deep UV resonance raman spectroscopy of early events of lysozyme fibrillation: kinetic mechanism and potential interpretation pitfalls
Boulet-Audet et al. High-throughput thermal stability analysis of a monoclonal antibody by attenuated total reflection FT-IR spectroscopic imaging
Minnes et al. Quantifying secondary structure changes in calmodulin using 2D-IR spectroscopy
Xu et al. The first step of hen egg white lysozyme fibrillation, irreversible partial unfolding, is a two‐state transition
Cunha et al. Assessing spectral simulation protocols for the amide I band of proteins
Shmool et al. An experimental approach probing the conformational transitions and energy landscape of antibodies: a glimmer of hope for reviving lost therapeutic candidates using ionic liquid
Hu et al. Investigation of protein conformational stability employing a multimodal spectrometer
Tajoddin et al. Analysis of temperature-dependent H/D exchange mass spectrometry experiments
Chalyavi et al. Tyrosine as a non-perturbing site-specific vibrational reporter for protein dynamics
Scheerer et al. Isotopically site-selected dynamics of a three-stranded β-sheet peptide detected with temperature-jump infrared-spectroscopy
Elliott et al. Comparative analysis of one-dimensional protein fingerprint by line shape enhancement and two-dimensional 1H, 13C methyl NMR methods for characterization of the higher order structure of IgG1 monoclonal antibodies
Knight et al. Monoclonal antibody stability can be usefully monitored using the excitation-energy-dependent fluorescence edge-shift
Fischer et al. Juxtaposition of Bub1 and Cdc20 on phosphorylated Mad1 during catalytic mitotic checkpoint complex assembly

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20180921

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

RIC1 Information provided on ipc code assigned before grant

Ipc: G01N 21/64 20060101AFI20190909BHEP

Ipc: G16C 20/30 20190101ALI20190909BHEP

Ipc: G16C 20/40 20190101ALI20190909BHEP

INTG Intention to grant announced

Effective date: 20191007

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

Ref country code: AT

Ref legal event code: REF

Ref document number: 1267584

Country of ref document: AT

Kind code of ref document: T

Effective date: 20200515

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602017016257

Country of ref document: DE

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: VALIPAT S.A. C/O BOVARD SA NEUCHATEL, CH

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200907

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200506

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200806

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200906

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200506

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200506

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200807

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2795997

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20201125

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200806

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200506

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200506

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200506

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 1267584

Country of ref document: AT

Kind code of ref document: T

Effective date: 20200506

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200506

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200506

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200506

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200506

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200506

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200506

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200506

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602017016257

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200506

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200506

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20210209

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200506

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200506

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210317

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200506

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230526

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20170317

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20230406

Year of fee payment: 7

Ref country code: CH

Payment date: 20230401

Year of fee payment: 7

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20240326

Year of fee payment: 8

Ref country code: IE

Payment date: 20240326

Year of fee payment: 8

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200506

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20240328

Year of fee payment: 8

Ref country code: GB

Payment date: 20240304

Year of fee payment: 8

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IT

Payment date: 20240322

Year of fee payment: 8

Ref country code: FR

Payment date: 20240327

Year of fee payment: 8

Ref country code: BE

Payment date: 20240326

Year of fee payment: 8

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200506